Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Nat Commun ; 15(1): 4711, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38830841

RESUMO

The fetal development of organs and functions is vulnerable to perturbation by maternal inflammation which may increase susceptibility to disorders after birth. Because it is not well understood how the placenta and fetus respond to acute lung- inflammation, we characterize the response to maternal pulmonary lipopolysaccharide exposure across 24 h in maternal and fetal organs using multi-omics, imaging and integrative analyses. Unlike maternal organs, which mount strong inflammatory immune responses, the placenta upregulates immuno-modulatory genes, in particular the IL-6 signaling suppressor Socs3. Similarly, we observe no immune response in the fetal liver, which instead displays metabolic changes, including increases in lipids containing docosahexaenoic acid, crucial for fetal brain development. The maternal liver and plasma display similar metabolic alterations, potentially increasing bioavailability of docosahexaenoic acid for the mother and fetus. Thus, our integrated temporal analysis shows that systemic inflammation in the mother leads to a metabolic perturbation in the fetus.


Assuntos
Feto , Lipopolissacarídeos , Fígado , Pulmão , Placenta , Feminino , Gravidez , Placenta/metabolismo , Placenta/imunologia , Animais , Feto/imunologia , Feto/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Fígado/metabolismo , Fígado/imunologia , Ácidos Docosa-Hexaenoicos/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/genética , Camundongos , Inflamação/imunologia , Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Adaptação Fisiológica/imunologia , Desenvolvimento Fetal/imunologia , Troca Materno-Fetal/imunologia , Interleucina-6/metabolismo , Interleucina-6/imunologia
2.
Front Immunol ; 13: 821542, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35185911

RESUMO

CD49a+ natural killer (NK) cells play a critical role in promoting fetal development and maintaining immune tolerance at the maternal-fetal interface during the early stages of pregnancy. However, given their residency in human tissue, thorough studies and clinical applications are difficult to perform. It is still unclear as to how functional human CD49a+ NK cells can be induced to benefit pregnancy outcomes. In this study, we established three no-feeder cell induction systems to induce human CD49a+ NK cells from umbilical cord blood hematopoietic stem cells (HSCs), bone marrow HSCs, and peripheral blood NK cells in vitro. These induced NK cells (iNKs) from three cell induction systems display high levels of CD49a, CD9, CD39, CD151 expression, low levels of CD16 expression, and no obvious cytotoxic capability. They are phenotypically and functionally similar to decidual NK cells. Furthermore, these iNKs display a high expression of growth-promoting factors and proangiogenic factors and can promote fetal growth and improve uterine artery blood flow in a murine pregnancy model in vivo. This research demonstrates the ability of human-induced CD49a+ NK cells to promote fetal growth via three cell induction systems, which could eventually be used to treat patients experiencing adverse pregnancy outcomes.


Assuntos
Desenvolvimento Fetal/imunologia , Integrina alfa1/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células Matadoras Naturais/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Integrina alfa1/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Camundongos , Gravidez
3.
Front Immunol ; 12: 771054, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745147

RESUMO

The placenta is a fetal-derived organ whose function is crucial for both maternal and fetal health. The human placenta contains a population of fetal macrophages termed Hofbauer cells. These macrophages play diverse roles, aiding in placental development, function and defence. The outer layer of the human placenta is formed by syncytiotrophoblast cells, that fuse to form the syncytium. Adhered to the syncytium at sites of damage, on the maternal side of the placenta, is a population of macrophages termed placenta associated maternal macrophages (PAMM1a). Here we discuss recent developments that have led to renewed insight into our understanding of the ontogeny, phenotype and function of placental macrophages. Finally, we discuss how the application of new technologies within placental research are helping us to further understand these cells.


Assuntos
Desenvolvimento Fetal/imunologia , Feto/imunologia , Imunidade Inata/imunologia , Macrófagos/imunologia , Placenta/imunologia , Animais , Movimento Celular/imunologia , Movimento Celular/fisiologia , Vilosidades Coriônicas/imunologia , Vilosidades Coriônicas/metabolismo , Feminino , Feto/citologia , Feto/fisiologia , Receptor 2 de Folato/imunologia , Receptor 2 de Folato/metabolismo , Antígenos HLA-DR/imunologia , Antígenos HLA-DR/metabolismo , Humanos , Macrófagos/metabolismo , Macrófagos/fisiologia , Fagocitose/imunologia , Fagocitose/fisiologia , Placenta/citologia , Placenta/fisiologia , Gravidez
4.
Br J Cancer ; 124(12): 1897-1899, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33767421
5.
Front Immunol ; 12: 637975, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33679795

RESUMO

Human B-lymphopoiesis is a dynamic life-long process that starts in utero by around six post-conception weeks. A detailed understanding of human fetal B-lymphopoiesis and how it changes in postnatal life is vital for building a complete picture of normal B-lymphoid development through ontogeny, and its relevance in disease. B-cell acute lymphoblastic leukemia (B-ALL) is one of the most common cancers in children, with many of the leukemia-initiating events originating in utero. It is likely that the biology of B-ALL, including leukemia initiation, maintenance and progression depends on the developmental stage and type of B-lymphoid cell in which it originates. This is particularly important for early life leukemias, where specific characteristics of fetal B-cells might be key to determining how the disease behaves, including response to treatment. These cellular, molecular and/or epigenetic features are likely to change with age in a cell intrinsic and/or microenvironment directed manner. Most of our understanding of fetal B-lymphopoiesis has been based on murine data, but many recent studies have focussed on characterizing human fetal B-cell development, including functional and molecular assays at a single cell level. In this mini-review we will give a short overview of the recent advances in the understanding of human fetal B-lymphopoiesis, including its relevance to infant/childhood leukemia, and highlight future questions in the field.


Assuntos
Linfócitos B/imunologia , Desenvolvimento Fetal/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Células Precursoras de Linfócitos B/imunologia , Carcinogênese , Diferenciação Celular , Humanos , Ativação Linfocitária
6.
Nat Rev Immunol ; 20(5): 279-293, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31853049

RESUMO

Much of our knowledge regarding the interactions between epithelial tissues and the immune system has been gathered from animal models and co-cultures with cell lines. However, unique features of human cells cannot be modelled in mice, and cell lines are often transformed or genetically immortalized. Organoid technology has emerged as a powerful tool to maintain epithelial cells in a near-native state. In this Review, we discuss how organoids are being used in immunological research to understand the role of epithelial cell-immune cell interactions in tissue development and homeostasis, as well as in diseases such as cancer.


Assuntos
Células Epiteliais/imunologia , Epitélio/imunologia , Desenvolvimento Fetal/imunologia , Infecções/imunologia , Linfopoese/imunologia , Organoides/imunologia , Regeneração/imunologia , Alergia e Imunologia , Pesquisa Biomédica , Diferenciação Celular/imunologia , Homeostase , Humanos , Linfócitos T/imunologia , Timo/imunologia , Microambiente Tumoral/imunologia
7.
Cell Mol Life Sci ; 76(4): 729-743, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30374520

RESUMO

Cannabinoids are the most commonly abused illicit drugs worldwide. While cannabis can be beneficial for certain heath conditions, abuse of potent synthetic cannabinoids has been on the rise. Exposure to cannabinoids is also prevalent in women of child-bearing age and pregnant women. These compounds can cross the placental barrier and directly affect the fetus. They mediate their effects primarily through G-protein coupled cannabinoid receptors, CB1 and CB2. In addition to significant neurological effects, cannabinoids can trigger robust immunomodulation by altering cytokine levels, causing apoptosis of lymphoid cells and inducing suppressor cells of the immune system. Profound effects of cannabinoids on the immune system as discussed in this review, suggest that maternal exposure during pregnancy could lead to dysregulation of innate and adaptive immune system of developing fetus and offspring potentially leading to weakening of immune defenses against infections and cancer later in life. Emerging evidence also indicates the underlying role of epigenetic mechanisms causing long-lasting impact following cannabinoid exposure in utero.


Assuntos
Canabinoides/intoxicação , Desenvolvimento Fetal/efeitos dos fármacos , Sistema Imunitário/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/imunologia , Imunidade Adaptativa/efeitos dos fármacos , Imunidade Adaptativa/imunologia , Canabinoides/química , Feminino , Desenvolvimento Fetal/imunologia , Humanos , Sistema Imunitário/embriologia , Sistema Imunitário/imunologia , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Estrutura Molecular , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente
8.
J Pathol ; 247(5): 686-696, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30506724

RESUMO

Lung development is a complex process mediated through the interaction of multiple cell types, factors and mediators. In mice, it starts as early as embryonic day 9 and continues into early adulthood. The process can be separated into five different developmental stages: embryonic, pseudoglandular, canalicular, saccular, and alveolar. Whilst lung bud formation and branching morphogenesis have been studied extensively, the mechanisms of alveolarisation are incompletely understood. Aberrant lung development can lead to deleterious consequences for respiratory health such as bronchopulmonary dysplasia (BPD), a disease primarily affecting preterm neonates, which is characterised by increased pulmonary inflammation and disturbed alveolarisation. While the deleterious effects of type 1-mediated inflammatory responses on lung development have been well established, the role of type 2 responses in postnatal lung development remains poorly understood. Recent studies indicate that type 2-associated immune cells, such as group 2 innate lymphoid cells and alveolar macrophages, are increased in number during postnatal alveolarisation. Here, we present the current state of understanding of the postnatal stages of lung development and the key cell types and mediators known to be involved. We also provide an overview of how stem cells are involved in lung development and regeneration, and the negative influences of respiratory infections. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Imunidade Adaptativa/fisiologia , Pulmão/crescimento & desenvolvimento , Angiotensina II/imunologia , Angiotensina II/fisiologia , Animais , Brônquios/citologia , Desenvolvimento Fetal/imunologia , Desenvolvimento Fetal/fisiologia , Humanos , Pulmão/embriologia , Pulmão/imunologia , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Neovascularização Fisiológica/imunologia , Neovascularização Fisiológica/fisiologia , Pneumonia/imunologia , Pneumonia/fisiopatologia , Regeneração/imunologia , Regeneração/fisiologia , Transdução de Sinais/imunologia , Transdução de Sinais/fisiologia
9.
Brain Behav Immun ; 70: 390-397, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29588230

RESUMO

Preclinical studies demonstrate that environmentally-induced alterations in inflammatory cytokines generated by the maternal and fetal immune system can significantly impact fetal brain development. Yet, the relationship between maternal cytokines during gestation and later cognitive ability and executive function remains understudied. Children (n = 246) were born of mothers enrolled in the Newborn Epigenetic Study - a prospective pre-birth cohort in the Southeastern US. We characterized seven cytokines [IL-1ß, IL-4,IL-6, IL-12p70, IL-17A, tumor necrosis factor-α (TNFα), and interferon-γ (IFNγ)] and one chemokine (IL-8) from maternal plasma collected during pregnancy. We assessed children's cognitive abilities and executive functioning at a mean age of 4.5 (SD = 1.1) years. Children's DAS-II and NIH toolbox scores were regressed on cytokines and the chemokine, controlling for maternal age, race, education, body mass index, IQ, parity, smoking status, delivery type, gestational weeks, and child birth weight and sex. Higher IL-12p70 (ßIL-12p70 = 4.26, p = 0.023) and IL-17A (ßIL-17A = 3.70, p = 0.042) levels were related to higher DAS-II GCA score, whereas higher IL-1ß (ßIL-1B = -6.07, p = 0.003) was related to lower GCA score. Higher IL-12p70 was related to higher performance on NIH toolbox measures of executive functions related to inhibitory control and attention (ßIL-12p70 = 5.20, p = 0.046) and cognitive flexibility (ßIL-12p70 = 5.10, p = 0.047). Results suggest that dysregulation in gestational immune activity are associated with child cognitive ability and executive functioning.


Assuntos
Cognição/fisiologia , Função Executiva/fisiologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Adulto , Peso ao Nascer , Índice de Massa Corporal , Quimiocinas/análise , Quimiocinas/sangue , Criança , Pré-Escolar , Cognição/efeitos dos fármacos , Estudos de Coortes , Citocinas/análise , Citocinas/sangue , Citocinas/metabolismo , Função Executiva/efeitos dos fármacos , Feminino , Desenvolvimento Fetal/imunologia , Desenvolvimento Fetal/fisiologia , Humanos , Recém-Nascido , Masculino , Mães , Gravidez , Estudos Prospectivos
10.
J Reprod Immunol ; 125: 80-88, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29306096

RESUMO

Cytokines in the reproductive tract environment at conception mediate a dialogue between the embryo and maternal tissues to profoundly influence embryo development and implantation success. Through effects on gene expression and the cell stress response, cytokines elicit an epigenetic impact with consequences for placental development and fetal growth, which in turn affect metabolic phenotype and long-term health of offspring. There is substantial evidence demonstrating that pro-survival cytokines, such as GM-CSF, CSF1, LIF, HB-EGF and IGFII, support embryos to develop optimally. Less attention has been paid to cytokines that adversely impact embryo development, including the pro-inflammatory cytokines TNF, TRAIL and IFNG. These agents elicit cell stress, impair cell survival and retard blastocyst development, and at sufficiently high concentrations, can cause embryo demise. Experiments in mice suggest these so-called 'embryotoxic' cytokines can harm embryos through pro-apoptotic and adverse programming effects, as well as indirectly suppressing uterine receptivity through the maternal immune response. Embryotrophic factors may mitigate against and protect from these adverse effects. Thus, the balance between embryotrophic and embryotoxic cytokines can impart effects on embryo development and implantation, and has the potential to contribute to endometrial 'biosensor' function to mediate embryo selection. Embryotoxic cytokines can be elevated in plasma and reproductive tract tissues in inflammatory conditions including infection, diabetes, obesity, PCOS and endometriosis. Studies are therefore warranted to investigate whether excessive embryotoxic cytokines contribute to infertility and recurrent implantation failure in women, and compromised reproductive performance in livestock animals.


Assuntos
Citocinas/imunologia , Perda do Embrião/imunologia , Desenvolvimento Embrionário/imunologia , Endométrio/imunologia , Desenvolvimento Fetal/imunologia , Animais , Citocinas/sangue , Citocinas/metabolismo , Perda do Embrião/sangue , Embrião de Mamíferos/imunologia , Endométrio/metabolismo , Feminino , Retardo do Crescimento Fetal/imunologia , Humanos , Infertilidade/imunologia , Doenças Metabólicas/imunologia , Placentação/imunologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/imunologia
11.
J Reprod Immunol ; 125: 45-55, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29253793

RESUMO

The preterm newborn is at significant risk of neural injury and impaired neurodevelopment. Infants with mild or no evidence of injury may also be at risk of altered brain development, with evidence impaired cell maturation. The underlying causes are multifactorial and include exposure of both the fetus and newborn to hypoxia-ischemia, inflammation (chorioamnionitis) and infection, adverse maternal lifestyle choices (smoking, drug and alcohol use, diet) and obesity, as well as the significant demand that adaptation to post-natal life places on immature organs. Further, many fetuses and infants may have combinations of these events, and repeated (multi-hit) events that may induce tolerance to injury or sensitize to greater injury. Currently there are no treatments to prevent preterm injury or impaired neurodevelopment. However, inflammation is a common pathway for many of these insults, and clinical and experimental evidence demonstrates that acute and chronic inflammation is associated with impaired brain development. This review examines our current knowledge about the relationship between inflammation and preterm brain development, and the potential for stem cell therapy to provide neuroprotection and neurorepair through reducing inflammation and release of trophic factors, which promote cell maturation and repair.


Assuntos
Encéfalo/embriologia , Hipóxia-Isquemia Encefálica/imunologia , Inflamação/imunologia , Transtornos do Neurodesenvolvimento/imunologia , Efeitos Tardios da Exposição Pré-Natal/imunologia , Animais , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/imunologia , Corioamnionite/imunologia , Modelos Animais de Doenças , Feminino , Desenvolvimento Fetal/imunologia , Feto/embriologia , Feto/imunologia , Humanos , Hipóxia-Isquemia Encefálica/embriologia , Recém-Nascido , Recém-Nascido Prematuro/crescimento & desenvolvimento , Recém-Nascido Prematuro/imunologia , Inflamação/embriologia , Oligodendroglia/imunologia , Gravidez
12.
Reprod Toxicol ; 74: 116-133, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28916434

RESUMO

MEDI-570 is a fully human afucosylated monoclonal antibody (MAb) against Inducible T-cell costimulator (ICOS), highly expressed on CD4+ T follicular helper (TFH) cells. Effects of MEDI-570 were evaluated in an enhanced pre-postnatal development toxicity (ePPND) study in cynomolgus monkeys. Administration to pregnant monkeys did not cause any abortifacient effects. Changes in hematology and peripheral blood T lymphocyte subsets in maternal animals and infants and the attenuated infant IgG immune response to keyhole limpet hemocyanin (KLH) were attributed to MEDI-570 pharmacology. Adverse findings included aggressive fibromatosis in one dam and two infant losses in the high dose group with anatomic pathology findings suggestive of atypical lymphoid hyperplasia. The margin of safety relative to the no observed adverse effect level (NOAEL) for the highest planned clinical dose in the Phase 1a study was 7. This study suggests that women of child bearing potential employ effective methods of contraception while being treated with MEDI-570.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Depleção Linfocítica , Linfócitos T/imunologia , Animais , Animais Recém-Nascidos , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Humanizados , Embrião de Mamíferos/imunologia , Desenvolvimento Embrionário/imunologia , Feminino , Desenvolvimento Fetal/imunologia , Feto/efeitos dos fármacos , Hemocianinas/farmacologia , Imunoglobulina G/imunologia , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Contagem de Linfócitos , Macaca fascicularis , Masculino , Troca Materno-Fetal , Gravidez
13.
J Immunol ; 196(2): 678-90, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26643476

RESUMO

Embryos and tumors are both masses of dividing cells expressing foreign Ags, but they are not rejected by the immune system. We hypothesized that similar tolerogenic mechanisms prevent their rejection. Global comparison of fetal and tumor microenvironments through transcriptomics in mice revealed strikingly similar and dramatic decreases in expression of numerous immune-related pathways, including Ag presentation and T cell signaling. Unsupervised analyses highlighted the parallel kinetics and similarities of immune signature downregulation, from the very first days after tumor or embryo implantation. Besides upregulated signatures related to cell proliferation, the only significant signatures shared by the two conditions across all biological processes and all time points studied were downmodulated immune response signatures. Regulatory T cell depletion completely reverses this immune downmodulation to an immune upregulation that leads to fetal or tumor immune rejection. We propose that evolutionarily selected mechanisms that protect mammalian fetuses from immune attack are hijacked to license tumor development.


Assuntos
Desenvolvimento Fetal/imunologia , Tolerância Imunológica/imunologia , Neoplasias/imunologia , Linfócitos T Reguladores/imunologia , Evasão Tumoral/imunologia , Animais , Feminino , Feto/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transcriptoma
14.
Morfologiia ; 150(4): 90-6, 2016.
Artigo em Russo | MEDLINE | ID: mdl-30137700

RESUMO

The problem of the formation of new lymphoid structures (neolymphogenesis) is quite controversial and widely discussed in the literature. Under normal conditions, lymphoid organs arise only in the process of fetal development (organogenesis), however in long-standing chronic inflammatory processes, nonhealing wounds, autoimmune diseases, oncologic pathology spontaneous formation of new lymphoid structures was noted. The structures of the peripheral lymphoid formations include the lymphocytes arranged singly and in clusters (infiltration), lymphoid nodules and lymph nodes. The morphogenesis of the components of lymphoid tissue and the possibility of creating artificial lymphoid structures, reproducing the function of the natural ones, is demonstrated. Important role in the development of lymphoid structures is played by mediators of inflammation, cytokines of the family of lymphotoxins, tumor necrosis factor. The possibilities of prosthetic substitution of the functions of the lymphoid structures are described for the activation of protective processes in the body.


Assuntos
Desenvolvimento Fetal/imunologia , Linfonodos , Linfócitos/imunologia , Linfotoxina-alfa/imunologia , Organogênese/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Humanos , Linfonodos/embriologia , Linfonodos/imunologia
17.
Immunity ; 43(2): 382-93, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26287683

RESUMO

Macrophages are one of the most diverse cell populations in terms of their anatomical location and functional specialization during both homeostasis and disease. Although it has been shown in different fate mapping models that some macrophages present in adult tissues are already established during fetal development, their exact origins are still under debate. In the current study, we developed a fate mapping strain, based on the Kit locus, which allowed us to readdress "the origins" question. Different types of macrophages from various adult tissues were traced to their fetal or adult sources by inducing labeling in precursors at several time points either during fetal development or in adult mice. We show that all adult macrophages, resident or infiltrating, are progenies of classical hematopoietic stem cells (HSC) with the exception of microglia and, partially epidermal Langerhans cells, which are yolk sac (YS)-derived.


Assuntos
Desenvolvimento Fetal/imunologia , Células-Tronco Hematopoéticas/fisiologia , Macrófagos/fisiologia , Microglia/fisiologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Animais , Antígenos CD/metabolismo , Diferenciação Celular , Linhagem da Célula , Embrião de Mamíferos , Feminino , Homeostase , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Proteínas Proto-Oncogênicas c-kit/genética , Saco Vitelino/fisiologia
19.
Cardiovasc Res ; 107(3): 352-63, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25987546

RESUMO

Fetal haematopoiesis is a highly regulated process in terms of time and location. It is characterized by the emergence of specific cell populations at different extra- and intraembryonic anatomical sites. Trafficking of haematopoietic stem cells (HSCs) between these supportive niches is regulated by a set of molecules, i.e. integrins and chemokine receptors, which are also described for the recruitment of differentiated innate immune cells. In this review, an overview will be given on fetal haematopoiesis as well as trafficking of HSCs during fetal life. In addition, we will focus on the appearance of the first differentiated neutrophils and monocytes in the fetal circulation and describe how they acquire the ability to roll, adhere, and transmigrate into inflamed fetal tissue. Furthermore, we will discuss other effector functions of innate immune cells evolving during fetal ontogeny.


Assuntos
Desenvolvimento Fetal/imunologia , Hematopoese , Células-Tronco Hematopoéticas/fisiologia , Monócitos/fisiologia , Neutrófilos/fisiologia , Animais , Humanos , Células Mieloides/fisiologia
20.
Eur J Immunol ; 45(1): 238-49, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25311115

RESUMO

Interleukin-1ß (IL-1ß) production is impaired in cord blood monocytes. However, the mechanism underlying this developmental attenuation remains unclear. Here, we analyzed the extent of variability within the Toll-like receptor (TLR)/NLRP3 inflammasome pathways in human neonates. We show that immature low CD14 expressing/CD16(pos) monocytes predominate before 33 weeks of gestation, and that these cells lack production of the pro-IL-1ß precursor protein upon LPS stimulation. In contrast, high levels of pro-IL-1ß are produced within high CD14 expressing monocytes, although these cells are unable to secrete mature IL-1ß. The lack of secreted IL-1ß in these monocytes parallels a reduction of NLRP3 induction following TLR stimulation resulting in a lack of caspase-1 activity before 29 weeks of gestation, whereas expression of the apoptosis-associated speck-like protein containing a CARD and function of the P2×7 receptor are preserved. Our analyses also reveal a strong inhibitory effect of placental infection on LPS/ATP-induced caspase-1 activity in cord blood monocytes. Lastly, secretion of IL-1ß in preterm neonates is restored to adult levels during the neonatal period, indicating rapid maturation of these responses after birth. Collectively, our data highlight important developmental mechanisms regulating IL-1ß responses early in gestation, in part due to a downregulation of TLR-mediated NLRP3 expression. Such mechanisms may serve to limit potentially damaging inflammatory responses in a developing fetus.


Assuntos
Proteínas de Transporte/imunologia , Desenvolvimento Fetal/imunologia , Inflamassomos/imunologia , Interleucina-1beta/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Trifosfato de Adenosina/farmacologia , Adulto , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Transporte/genética , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/imunologia , Sangue Fetal/citologia , Sangue Fetal/efeitos dos fármacos , Sangue Fetal/imunologia , Feto , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Inflamassomos/genética , Interleucina-1beta/genética , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Monócitos/citologia , Monócitos/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/imunologia , Transdução de Sinais , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA